US20110159092A1 - Extended release pharmaceutical composition comprising linezolid and process for preparing the same - Google Patents

Extended release pharmaceutical composition comprising linezolid and process for preparing the same Download PDF

Info

Publication number
US20110159092A1
US20110159092A1 US12/980,776 US98077610A US2011159092A1 US 20110159092 A1 US20110159092 A1 US 20110159092A1 US 98077610 A US98077610 A US 98077610A US 2011159092 A1 US2011159092 A1 US 2011159092A1
Authority
US
United States
Prior art keywords
linezolid
pharmaceutical composition
extended release
poly
composition according
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/980,776
Inventor
Rajesh Kshirsagar
Sachin Mundade
Ganesh SHINDE
Pravin KAMBLE
SM Mudda
Shivanand DHANURE
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Micro Labs Ltd
Original Assignee
Micro Labs Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Micro Labs Ltd filed Critical Micro Labs Ltd
Assigned to MICRO LABS LIMITED reassignment MICRO LABS LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DHANURE, SHIVANAND, KAMBLE, PRAVIN, KSHIRSAGAR, RAJESH, MUDDA, SM, MUNDADE, SACHIN, SHINDE, GANESH
Publication of US20110159092A1 publication Critical patent/US20110159092A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2027Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2031Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2086Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat
    • A61K9/209Layered tablets, e.g. bilayer tablets; Tablets of the type inert core-active coat containing drug in at least two layers or in the core and in at least one outer layer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds
    • A61K9/286Polysaccharides, e.g. gums; Cyclodextrin
    • A61K9/2866Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents

Definitions

  • the present invention relates to an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients and a process for preparing the same.
  • Linezolid is one of the oxazolidinone antibiotics with a chemical name (S)-N-( ⁇ 3-[3-fluoro-4-(morpholin-4-yl)phenyl]-2-oxo-1,3-oxazolidin-5-yl ⁇ methyl)acetamide it is orally effective and highly soluble in water.
  • Linezolid has clinical utility in the treatment of infections caused by aerobic Gram-positive bacteria.
  • the in vitro spectrum of activity of Linezolid also includes certain Gram-negative bacteria and anaerobic bacteria.
  • Linezolid inhibits bacterial protein synthesis through a mechanism of action different from that of other antibacterial agents, therefore, cross-resistance between Linezolid and other classes of antibiotics is unlikely.
  • Linezolid binds to a site on the bacterial 23S ribosomal RNA of the 50S subunit and prevents the formation of a functional 70S initiation complex, which is an essential component of the bacterial translation process.
  • the results of time-kill studies have shown Linezolid to be bacteriostatic against enterococci and staphylococci . For streptococci , Linezolid was found to be bactericidal for the majority of strains.
  • Linezolid is commercially sold as immediate release (IR) tablet under the trade name Zyvox®. These tablets for oral administration contain 400 mg or 600 mg Linezolid as film-coated compressed tablets and also contain corn starch, microcrystalline cellulose, hydroxypropylcellulose, sodium starch glycolate, magnesium stearate, hypromellose, polyethylene glycol, titanium dioxide, and carnauba wax. The drug appears to inhibit the initiation of protein synthesis at the ribosomal level in susceptible bacteria.
  • IR immediate release
  • Linezolid belongs to antibiotics category which requires blood concentration of drug to be maintained above MIC (Minimum inhibitory concentration) level for longer period of time to have effective antibacterial activity.
  • Linezolid is prescribed for the treatment of infections like community acquired pneumonia, including concurrent bacteremia, complicated skin and skin structure infections, nosocomial pneumonia at 10 to 14 days recommended duration of treatment, vancomycin resistant enterococcus faecium infections including concurrent bacteremia at 14 to 28 days recommended duration of treatment.
  • U.S. Pat. No. 6,514,529 (“the '529 patent”) discloses immediate release tablets containing a high drug content of oxazolidinones such as Linezolid, wherein the oxazolidinone is compressed into a tablet using corn starch, microcrystalline cellulose, hydroxylpropyl cellulose, sodium starch glycolate and magnesium stearate as the excipients.
  • the '529 patent further discloses method for providing blood levels of Linezolid by oral administration medically equivalent to the blood levels produced by IV administration of the Linezolid which comprises administration of immediate release tablet formulation.
  • U.S. Pat. No. 6,451,345 disclose taste masked microcapsule compositions for oxazolidinone or macrolide antibiotics comprise microcapsules of drug coated by coacervation of a microencapsulation polymer and a coated plasticized enteric polymer.
  • Extended or sustained or slow release compositions offer clinically significant advantages for various therapeutically active agents by way of increasing patient compliance due to reduced frequency of administration; improve the safety and efficacy of drug substances and reduce undesirable effects in comparison to the corresponding immediate release dosage form.
  • immediate-release compositions When immediate-release compositions are given higher peak plasma concentrations are achieved this sometimes may be undesirable due to toxic effects.
  • extended release formulations releases the drug over prolonged period of time in controlled release manner the plasma concentrations may be achieved and maintained which are below toxic levels.
  • Immediate release compositions in case of antibiotics due to the shorter half life eliminates faster from body and the plasma concentration drop down below MIC level and may not be effective; therefore for immediate release formulations the frequency of dosing required is more.
  • the in-vivo plasma concentrations are maintained above MIC level for longer period of time therefore reducing dosing frequency and improving efficacy which is desirable especially for antibiotics.
  • Linezolid belongs to antibiotics or antibacterial category which requires high dose of drug frequently (BID) to achieve required MIC level.
  • BID drug frequently
  • T>MIC time above minimum inhibitory concentration
  • C max maximum plasma concentration
  • Linezolid is high dose (1200/day) and high solubility (3 mg/ml) active. These types of actives pose challenges to a formulator as it is difficult to formulate an extended release dosage form which can be suitable for once daily dosing for such actives because:
  • an object of the present invention to provide an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • an extended release solid pharmaceutical composition suitable for once daily dosing such as tablet comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • a further aspect of the present invention provides a process for the preparation of an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • FIG. 1 shows dissolution profiles of the pharmaceutical compositions according to the present invention for Example 1 to Example 19 using USP Type II (Paddle Apparatus) at 50 rpm in 900 ml/1000 ml, 0.01N HCl for first 2 hours, followed by the media with pH 6.8 Phosphate buffer.
  • USP Type II Phase Apparatus
  • FIG. 2 shows comparative dissolution profiles of the pharmaceutical compositions according to the present invention for Example 11 and Example 15 using USP Type II (Paddle Apparatus) at 50 rpm in 900 ml/1000 ml, 0.1N HCl, pH 4.5 Acetate buffer and pH 6.8 Phosphate buffer.
  • USP Type II Phase Apparatus
  • FIG. 3 shows comparative mean plasma concentration ( ⁇ g/ml) vs. time (hours) profile for 1200 mg (600 mg ⁇ 2 Tablets), 1000 mg ER tablets according to Example 11 (Test 1) and Example 15 (Test 2) respectively and reference immediate release Zyvox 600 mg tablet twice daily.
  • Linezolid refers to any pharmaceutically acceptable form of Linezolid including base or its pharmaceutically acceptable complexes, salts, prodrug, derivative polymorphs, hydrates, solvates, enantiomers or racemates, metabolite as dictated by the context of its use.
  • Linezolid may be present from about 50 mg to about 1700 mg Linezolid; preferably from about 300 mg to about 1500 mg Linezolid, preferably about 600 mg, about 1000 mg, about 1200 mg or about 1500 mg of Linezolid.
  • Extended release pharmaceutical composition refers to any composition or dosage form that comprises an active drug and which is formulated to provide a longer duration of pharmacological response after administration of the dosage form than is ordinarily experienced after administration of a corresponding immediate release composition.
  • Extended release compositions include, inter alia, those compositions described elsewhere as “controlled release”, “delayed release”, “sustained release”, “prolonged release”, “programmed release”, “time release” and/or “rate controlled” compositions or dosage forms.
  • pharmaceutically acceptable is meant a carrier comprised of a material that is not biologically or otherwise undesirable.
  • C max means maximum plasma concentration of Linezolid, produced by the oral administration of the composition of the invention or the immediate release (IR) comparator.
  • T max means time to the maximum observed plasma concentration.
  • AUC 0-24 as used herein, means area under the plasma concentration-time curve, as calculated by the trapezoidal rule over the complete 24-hour interval for all the formulations.
  • T>MIC means time above MIC (minimum inhibitory concentration) calculated manually by graphical interpolation, where the minimum inhibitory plasma concentration was defined as 2 ⁇ g/ml of Linezolid.
  • “Adverse effects” as used herein, means those physiological effects to various systems in the body such as cardiovascular systems, nervous system, digestive system, and body as a whole, which cause pain and discomfort to the individual subject.
  • the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • the present invention provides an extended release tablet suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • the present invention provides an extended release matrix tablet suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • the present invention provides an extended release coated tablet suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • the present invention provides a process of preparing an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more release controlling material(s).
  • the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more hydrophilic release controlling material(s).
  • the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more hydrophobic release controlling material(s).
  • the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and a matrix comprising one or more hydrophilic release controlling material(s) optionally coated with hydrophilic material and/or hydrophobic material or combinations thereof.
  • the present invention provides an extended release tablet suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more hydrophilic release controlling material(s) optionally coated with hydrophilic material and/or hydrophobic material or combinations thereof.
  • the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and pharmaceutically acceptable excipient having a in-vitro dissolution rate when measured using the USP Type II (Paddle apparatus) at 50 rpm in 900 ml/1000 ml, 0.01N hydrochloric acid for first 2 hours followed by the media with pH 6.8 phosphate buffer at 37° C.
  • the present invention provides an extended release tablet comprising Linezolid having two layers comprising immediate release layer and controlled release layer; wherein immediate release layer comprises of loading dose and controlled release layer comprises of maintenance dose wherein loading dose is released immediately and maintenance dose released in controlled manner over a period of time.
  • the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients so that upon oral administration the maximum concentrations (C max ) of Linezolid in plasma are statistically significantly lower than the immediate release formulation given twice daily, and area under the plasma concentration-time curve (AUC) and the minimum plasma concentration are maintained over 24 hours.
  • C max maximum concentrations of Linezolid in plasma are statistically significantly lower than the immediate release formulation given twice daily, and area under the plasma concentration-time curve (AUC) and the minimum plasma concentration are maintained over 24 hours.
  • the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients so that upon oral administration, the composition induces lower fluctuation in the mean plasma concentration than an immediate release composition of the Linezolid.
  • the present invention provides a method of using an extended release, pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof as an active ingredient, and one or more pharmaceutically acceptable excipients, comprising administering the composition in an effective amount for the treatment for bacterial infection in a mammal.
  • the present invention provides a method of treating bacterial infections in a mammal comprising administering an extended release, pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof as an active ingredient, and one or more pharmaceutically acceptable excipients, capable of maintaining a serum level of the Linezolid above MIC level (2 ⁇ g/ml) for at least 24 hours.
  • the present invention provides method of treating bacterial infections in a mammal comprising administering an extended release, pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof as an active ingredient, and one or more pharmaceutically acceptable excipients, capable of maintaining T>MIC for at least 24 hours.
  • an extended release pharmaceutical composition according to present invention comprises of Linezolid from about 10% w/w to about 95% w/w of the composition, preferably from about 50% w/w to about 95% w/w of the composition.
  • an extended release pharmaceutical compositions of present invention upon oral administration to a human provides a mean maximum plasma concentration (C max ) of Linezolid not more than, about 16 ⁇ g/ml.
  • the active ingredient in the preparation according to the invention may suitably be incorporated in a matrix.
  • a matrix This may be any matrix that affords extended release of Linezolid that affords in-vitro dissolution rates over 24 hours.
  • the matrix is an extended release matrix.
  • immediate release matrices having a release controlling coating or controlled release matrices having immediate release coating, which provides extended release of the active ingredient may be used.
  • Tableting is preferred production method because it is faster, easier, adds fewer steps to the process and is the most economical. Further, the tableting method ensures a high production yield, contrary to the manufacture of pellets where the loss of production output is usually much higher. Excipients for the formulation were chosen carefully to give appropriate dissolution rate and stability of the finished dosage form.
  • the present invention provides, a process for preparing an extended release pharmaceutical composition
  • an extended release pharmaceutical composition comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, and metabolite thereof and water soluble binder
  • process can be selected from direct compression, dry granulation, wet granulation (aqueous/non-aqueous or combination) and melt granulation.
  • an extended release composition according to present invention comprises of one or more release controlling materials.
  • the release controlling materials can be hydrophilic release controlling materials or hydrophobic release controlling materials.
  • the release controlling materials are present from about 0.1% to about 20% w/w of the composition.
  • hydrophilic release controlling materials include but are not limited to hydroxypropyl methylcellulose, hydroxypropyl cellulose, methylcellulose, povidone, polyethylene glycols, vinyl acetate copolymers, polysaccharides such as alginates, xanthan gum, chitosan, carrageenan, dextran and the like, polyalkylene oxides such as polyethylene oxide and the likes, methacrylic acid copolymers, maleic anhydride/methyl vinyl ether copolymers, carbomers and the like.
  • hydrophobic release controlling materials include but are not limited to polyvinyl acetate dispersion, ethyl cellulose, cellulose acetate, cellulose propionate (lower, medium or higher molecular weight), cellulose acetate propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose triacetate, poly (methyl methacrylate), poly (ethyl methacrylate), poly (butyl methacrylate), poly (isobutyl methacrylate), and poly (hexyl methacrylate), poly (isodecyl methacrylate), poly (lauryl methacrylate), poly (phenyl methacrylate), poly (methyl acrylate), poly (isopropyl acrylate), poly (isobutyl acrylate), poly (octadecyl acrylate), waxes such as beeswax, carnauba wax, paraffin wax, microcrystalline wax, ozokerite; fatty
  • the formulation will, in general comprise of one or more excipients.
  • excipients include, but are not limited to, diluents, disintegrants, lubricant, glidant, binders, fillers, surfactant, solubilizers, wetting agents, chelating agents, stabilizers, alkalizing agents or amino acids.
  • a combination of excipients may also be used.
  • the amount of excipient(s) employed will depend upon how much active agent is to be used. One excipient can perform more than one function.
  • Binders include, but are not limited to, starches such as potato starch, wheat starch, corn starch; microcrystalline cellulose such as products known under the registered trade marks Avicel, Filtrak, Heweten or Pharmacel; celluloses such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxypropylmethyl cellulose (HPMC), ethyl cellulose, sodium carboxy methyl cellulose; natural gums like acacia, alginic acid, guar gum; liquid glucose, dextrin, povidone, syrup, polyethylene oxide, polyvinyl pyrrolidone, poly-N-vinyl amide, polyethylene glycol, gelatin, poly propylene glycol, tragacanth, combinations there of and other materials known to one of ordinary skill in the art.
  • starches such as potato starch, wheat starch, corn starch
  • microcrystalline cellulose such as products known under the registered trade marks Avicel, Filtrak, Heweten or Pharmacel
  • celluloses such as hydroxy
  • Fillers or diluents which include, but are not limited to confectioner's sugar, compressible sugar, dextrates, dextrin, dextrose, fructose, lactitol, mannitol, sucrose, starch, lactose, xylitol, sorbitol, talc, microcrystalline cellulose, calcium carbonate, calcium phosphate dibasic or tribasic, calcium sulphate, and the like can be used.
  • Lubricants may be selected from, but are not limited to, those conventionally known in the art such as Mg, Al, Ca or Zn stearate, polyethylene glycol, glyceryl behenate, mineral oil, sodium stearyl fumarate, stearic acid, hydrogenated vegetable oil and talc.
  • Glidants include, but are not limited to, silicon dioxide, magnesium trisilicate, powdered cellulose, starch, talc and tribasic calcium phosphate, calcium silicate, magnesium silicate, colloidal silicon dioxide, silicon hydrogel and other materials known to one of ordinary skill in the art.
  • the pharmaceutical composition according to the present invention include but is not limited to tablets (single layered tablets, multilayered tablets, mini tablets, bioadhesive tablets, caplets, matrix tablets, tablet within a tablet, mucoadhesive tablets, modified release tablets, pulsatile release tablets, timed release tablets), pellets, beads, granules, sustained release formulations, capsules, microcapsules, tablets in capsules and microspheres, matrix formulations, microencapsulation and powder/pellets/granules for suspension.
  • tablets single layered tablets, multilayered tablets, mini tablets, bioadhesive tablets, caplets, matrix tablets, tablet within a tablet, mucoadhesive tablets, modified release tablets, pulsatile release tablets, timed release tablets
  • pellets beads, granules, sustained release formulations, capsules, microcapsules, tablets in capsules and microspheres, matrix formulations, microencapsulation and powder/pellets/granules for suspension.
  • the pharmaceutical composition of the invention can optionally have one or more coatings such as film coating, sugar coating, enteric coating, bioadhesive coating and other coatings known in the art. These coatings help pharmaceutical formulations to release the drug at the required site of action.
  • coatings such as film coating, sugar coating, enteric coating, bioadhesive coating and other coatings known in the art. These coatings help pharmaceutical formulations to release the drug at the required site of action.
  • the additional coating prevents the dosage form from contacting the mouth or esophagus.
  • the additional coating remains intact until reaching the small intestine (e.g., an enteric coating).
  • Premature exposure of a bioadhesive layer or dissolution of a pharmaceutical dosage form in the mouth can be prevented with a layer or coating of hydrophilic polymers such as HPMC or gelatin.
  • Eudragit FS 30D or other suitable polymer may be incorporated in coating composition to retard the release of the drug to ensure drug release in the colon.
  • These coating layers comprises one or more excipients selected from the group comprising coating agents, opacifiers, taste-masking agents, fillers, polishing agents, colouring agents, antitacking agents and the like.
  • Coating agents which are useful in the coating process, include, but are not limited to, polysaccharides such as maltodextrin, alkyl celluloses such as methyl or ethyl cellulose, hydroxyalkylcelluloses (e.g. hydroxypropylcellulose or hydroxypropylmethylcelluloses); polyvinylpyrrolidone, acacia, corn, sucrose, gelatin, shellac, cellulose acetate pthalate, lipids, synthetic resins, acrylic polymers, opadry, polyvinyl alcohol (PVA), copolymers of vinylpyrrolidone and vinyl acetate (e.g.
  • PVA polyvinyl alcohol
  • additives can include plasticizers such as dibutyl phthalate, triethyl citrate, polyethylene glycol (PEG) and the like, antitacking agents such as talc, stearic acid, magnesium stearate and colloidal silicon dioxide and the like, surfactants such as polysorbates and sodium lauryl sulphate, fillers such as talc, precipitated calcium carbonate, polishing agents such as beeswax, carnauba wax, synthetic chlorinated wax and opacifying agents such as titanium dioxide and the like. All these excipients can be used at levels well known to the persons skilled in the art.
  • plasticizers such as dibutyl phthalate, triethyl citrate, polyethylene glycol (PEG) and the like
  • antitacking agents such as talc, stearic acid, magnesium stearate and colloidal silicon dioxide and the like
  • surfactants such as polysorbates and sodium lauryl sulphate
  • fillers such as talc, precipit
  • composition of the invention can be coated by a wide variety of methods. Suitable methods include compression coating, coating in a fluidized bed or a pan and hot melt (extrusion) coating. Such methods are well known to those skilled in the art.
  • Non-permeable coatings of insoluble polymers e.g., cellulose acetate, ethylcellulose
  • enteric coatings for delayed/modified release DR/MR
  • soluble pore formers e.g., PEG, PVA, sugars, salts, detergents, triethyl citrate, triacetin, etc.
  • Multi-layer or gradient tablets can be assembled in several different ways.
  • Linezolid and microcrystalline cellulose were sifted through suitable sieve and mixed.
  • the blend was granulated with Eudragit NE 30D and sufficient quantity of purified water.
  • the granules were dried and sifted through suitable sieve.
  • the Magnesium Stearate was sifted through suitable sieve and mixed with granules.
  • the blend was compressed into tablets.
  • Linezolid, lactose monohydrate and povidone were sifted through suitable sieve and mixed.
  • the blend was granulated with sufficient quantity of purified water.
  • the granules were dried and sifted through suitable sieve.
  • the Magnesium Stearate was sifted through suitable sieve and mixed with granules.
  • the blend was compressed into tablets.
  • Linezolid, lactose monohydrate and povidone were sifted through suitable sieve and mixed.
  • the blend was granulated with sufficient quantity of purified water.
  • the granules were dried and sifted through suitable sieve.
  • the Magnesium Stearate was sifted through suitable sieve and mixed with granules.
  • the blend was compressed into tablets.
  • Linezolid, lactose monohydrate and povidone were sifted through suitable sieve and mixed.
  • the blend was granulated with sufficient quantity of purified water.
  • the granules were dried and sifted through suitable sieve.
  • the Magnesium Stearate was sifted through suitable sieve and mixed with granules.
  • the blend was compressed into tablets.
  • Linezolid, lactose monohydrate and povidone were sifted through suitable sieve and mixed.
  • the blend was granulated with sufficient quantity of purified water.
  • the granules were dried and sifted through suitable sieve.
  • the Magnesium Stearate was sifted through suitable sieve and mixed with granules.
  • the blend was compressed into tablets.
  • Linezolid, Polyethylene Glycol 6000, lactose monohydrate and povidone were sifted through suitable sieve and mixed.
  • the blend was granulated with sufficient quantity of purified water.
  • the granules were dried and sifted through suitable sieve.
  • the Magnesium Stearate was sifted through suitable sieve and mixed with granules.
  • the blend was compressed into tablets.
  • Linezolid, lactose monohydrate and ethylcellulose were sifted through suitable sieve and mixed.
  • the povidone was dissolved in sufficient quantity of purified water.
  • the blend was granulated using povidone solution.
  • the granules were dried and sifted through suitable sieve.
  • the Magnesium Stearate was sifted through suitable sieve and mixed with granules.
  • the blend was compressed into tablets.
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried and sifted.
  • Talc and magnesium stearate were sifted and mixed with granules.
  • the blend was compressed into tablets.
  • Linezolid, corn starch, microcrystalline cellulose and hydroxypropylcellulose were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate.
  • the blend was mixed with sifted talc and magnesium stearate.
  • Linezolid, lactose monohydrate and povidone were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried and sifted.
  • the magnesium stearate was sifted and mixed with granules.
  • the blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablets.
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried and sifted.
  • Talc and magnesium stearate was sifted and mixed with granules.
  • the blend was compressed into tablets.
  • Linezolid, microcrystalline cellulose, hypromellose and Carboxymethylcellulose Calcium were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate.
  • the blend was mixed with sifted talc and magnesium stearate.
  • Linezolid, lactose monohydrate and povidone were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried and sifted.
  • the granules were mixed with sifted talc and magnesium stearate.
  • the blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablet.
  • Linezolid, microcrystalline cellulose, hypromellose and Carboxymethylcellulose Calcium were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate.
  • the blend was mixed with sifted talc and magnesium stearate.
  • Linezolid, lactose monohydrate and povidone were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried and sifted.
  • the granules were mixed with sifted talc and magnesium stearate.
  • the blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablet.
  • Controlled Release Inner Core Linezolid 450.0 Lactose monohydrate 73.0 Povidone 15.0 Purified Water q.s. Talc 6.0 Magnesium Stearate 6.0 Total Controlled Release Core 550.0 Immediate Release outer compression Coat Linezolid 150.0 Microcrystalline Cellulose 500.0 Carboxymethylcellulose 60.0 Calcium Hypromellose 15.0 Purified Water q.s. Microcrystalline Cellulose 210.0 Sodium Starch Glycolate 45.0 Talc 10.0 Magnesium Stearate 10.0 Total 1000.0 Total weight of compression 1550.0 coated tablet
  • Linezolid, lactose monohydrate and povidone were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried and sifted.
  • the granules were mixed with sifted talc and magnesium stearate.
  • the blend was compressed into tablet.
  • Linezolid, microcrystalline cellulose, hypromellose and Carboxymethylcellulose Calcium were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules was dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate.
  • the blend was mixed with sifted talc and magnesium stearate.
  • the inner core tablets were compression coated with the immediate release blend to form a controlled release tablet inside the immediate release tablets.
  • Linezolid, microcrystalline cellulose, hypromellose and Carboxymethylcellulose Calcium were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate.
  • the blend was mixed with sifted talc and magnesium stearate.
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried and sifted.
  • the granules were mixed with sifted talc and magnesium stearate.
  • the blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablet.
  • Linezolid, microcrystalline cellulose, hypromellose and Carboxymethylcellulose Calcium were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate.
  • the blend was mixed with sifted talc and magnesium stearate.
  • Linezolid, lactose monohydrate and povidone were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried and sifted.
  • the granules were mixed with sifted talc and magnesium stearate.
  • the blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablet.
  • Linezolid, microcrystalline cellulose and HPMC were sifted and mixed.
  • the blend was granulated with purified water.
  • the granules were dried, sifted and mixed with sifted talc and magnesium stearate.
  • the blend was compressed into tablets.
  • Linezolid, dibasic calcium phosphate, croscarmellose sodium and carboxymethylcellulose calcium were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried, sifted and mixed with sifted croscarmellose sodium.
  • the blend was mixed with sifted magnesium stearate.
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried and sifted.
  • the granules were mixed with sifted talc and magnesium stearate.
  • the blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablets.
  • Linezolid, dibasic calcium phosphate, croscarmellose sodium and carboxymethylcellulose calcium was sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules was dried, sifted and mixed with sifted croscarmellose sodium.
  • the blend was mixed with sifted magnesium stearate.
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried and sifted.
  • the granules were mixed with sifted talc and magnesium stearate.
  • the blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablets.
  • Linezolid, dibasic calcium phosphate, croscarmellose sodium and carboxymethylcellulose calcium were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried, sifted and mixed with sifted croscarmellose sodium.
  • the blend was mixed with sifted magnesium stearate.
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed.
  • the blend was granulated using sufficient quantity of purified water.
  • the granules were dried and sifted.
  • the granules were mixed with sifted talc and magnesium stearate.
  • the blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablets.
  • Linezolid, corn starch, microcrystalline cellulose and hydroxypropylcellulose were sifted and mixed.
  • the blend was granulated with sufficient quantity of purified water.
  • the granules were dried, sifted and mixed with microcrystalline cellulose and sodium starch glycolate.
  • the blend was mixed with magnesium stearate and compressed into tablets.
  • HPMC based coating material (Opadry Clear) was dissolved in sufficient quantity of purified water and mixed with ethylcellulose based aqueous coating material (Surelease). The core tablets were coated with the coating solution to a desired weight gain.
  • Linezolid, corn starch, microcrystalline cellulose and hydroxypropylcellulose were sifted and mixed.
  • the blend was granulated with sufficient quantity of purified water.
  • the granules were dried, sifted and mixed with microcrystalline cellulose and sodium starch glycolate.
  • the blend was mixed with magnesium stearate and compressed into tablets.
  • the pharmacokinetic study to determine the concentration-time plasma profile was done on healthy male subjects.
  • the study was conducted as open label, balanced, randomized, three-treatment, single-period, parallel, comparative oral bioavailability study as described below:
  • the study was conducted according to open label, balanced, randomized, three-treatment, single-period, parallel, comparative oral bioavailability study.
  • Plasma samples were analyzed to quantify the concentrations of Linezolid using a validated LC/MS/MS bioanalytical method.
  • Example No. 1 to Example No. 9 The in vitro dissolution is carried out using USP Type II (Paddle Apparatus) at 50 rpm in 900 ml/1000 ml, 0.01N HCl first two hours, followed by the media with pH 6.8 Phosphate buffer. The release of drug is measured using UV techniques.
  • USP Type II Phase Apparatus
  • the dissolution pattern for Example No. 1 to Example No. 9 is given in the table below
  • Dissolution Dissolution Media 900 ml/1000 ml 0.01N HCl for first 2 hours followed by media pH 6.8 Phosphate Buffer, USP II, 50 RPM Example No. 1 2 3 4 5 6 7 8 9 Time (Hours) Cumulative % Drug Released 1 41 10 9 9 13 11 9 10 33 2 67 18 17 17 25 19 13 18 39 4 96 32 29 28 42 33 22 37 48 6 100 40 38 37 55 43 30 54 54 8 — 49 46 45 66 54 38 83 59 10 — 58 53 52 76 62 45 85 64 12 — 66 59 58 84 69 53 107 69 14 — 71 65 64 92 76 59 — 74 16 — 79 70 70 99 82 67 — 79 20 — 91 80 80 — 93 78 — 87 24 — 101 89 89 — 101 88 — 92
  • Example No. 10 to Example No. 19 The in vitro dissolution is carried out using USP Type II (Paddle Apparatus) at 50 rpm in 900 ml/1000 ml, 0.01N HCl first two hours, followed by the media with pH 6.8 Phosphate buffer. The release of drug is measured using UV techniques.
  • the dissolution pattern for Example No. 10 to Example No. 19 is given in the table below:
  • Dissolution Dissolution Media 900 ml/1000 ml, 0.01N HCl for first 2 hours followed by media pH 6.8 Phosphate Buffer, USP II, 50 RPM Example 10 11 12 13 14 15 16 17 18 19 Time (hours) Cumulative % Drug Released 1 43 33 30 32 30 30 6 35 34 33 2 68 39 35 34 37 36 11 42 40 41 4 91 49 43 45 51 45 22 59 50 51 6 94 55 49 53 66 52 27 75 60 60 8 105 60 54 61 82 58 33 89 71 70 10 — 66 59 67 93 63 41 98 81 78 12 — 71 63 73 98 68 50 101 88 86 14 — 75 66 78 100 71 57 — 95 92 16 — 80 69 83 — 76 64 — 100 98 20 — 88 75 91 — 80 73 — — 24 — 95 80 97 — 84 85 — — — — — —
  • Example No. 11 and Example No. 15 The in vitro dissolution is carried out using USP Type II (Paddle Apparatus) at 50 rpm in 900 ml/1000 ml, 0.1 N HCl, pH 4.5 Acetate Buffer and pH 6.8 Phosphate Buffer. The release of drug is measured using UV techniques.
  • the dissolution pattern for Example No. 11 and Example No. 15 is given in the table below:

Abstract

The present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients and a process of preparing the same. The present invention further provides a method of treating bacterial infections in a mammal comprising administering an extended release, pharmaceutical composition suitable for once daily dosing comprising Linezolid capable of maintaining T>MIC for at least 24 hours. The present invention further provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid so that upon oral administration the maximum concentrations (Cmax) of Linezolid in plasma are statistically significantly lower than the immediate release formulation given twice daily, and area under the plasma concentration-time curve (AUC) and the minimum plasma concentrations are maintained over 24 hours.

Description

    FIELD OF THE INVENTION
  • The present invention relates to an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients and a process for preparing the same.
  • BACKGROUND OF THE INVENTION
  • Linezolid is one of the oxazolidinone antibiotics with a chemical name (S)-N-({3-[3-fluoro-4-(morpholin-4-yl)phenyl]-2-oxo-1,3-oxazolidin-5-yl}methyl)acetamide it is orally effective and highly soluble in water.
  • Linezolid has clinical utility in the treatment of infections caused by aerobic Gram-positive bacteria. The in vitro spectrum of activity of Linezolid also includes certain Gram-negative bacteria and anaerobic bacteria. Linezolid inhibits bacterial protein synthesis through a mechanism of action different from that of other antibacterial agents, therefore, cross-resistance between Linezolid and other classes of antibiotics is unlikely. Linezolid binds to a site on the bacterial 23S ribosomal RNA of the 50S subunit and prevents the formation of a functional 70S initiation complex, which is an essential component of the bacterial translation process. The results of time-kill studies have shown Linezolid to be bacteriostatic against enterococci and staphylococci. For streptococci, Linezolid was found to be bactericidal for the majority of strains.
  • Linezolid is commercially sold as immediate release (IR) tablet under the trade name Zyvox®. These tablets for oral administration contain 400 mg or 600 mg Linezolid as film-coated compressed tablets and also contain corn starch, microcrystalline cellulose, hydroxypropylcellulose, sodium starch glycolate, magnesium stearate, hypromellose, polyethylene glycol, titanium dioxide, and carnauba wax. The drug appears to inhibit the initiation of protein synthesis at the ribosomal level in susceptible bacteria.
  • Linezolid belongs to antibiotics category which requires blood concentration of drug to be maintained above MIC (Minimum inhibitory concentration) level for longer period of time to have effective antibacterial activity. Linezolid is prescribed for the treatment of infections like community acquired pneumonia, including concurrent bacteremia, complicated skin and skin structure infections, nosocomial pneumonia at 10 to 14 days recommended duration of treatment, vancomycin resistant enterococcus faecium infections including concurrent bacteremia at 14 to 28 days recommended duration of treatment. This longer duration of treatment exposes the patient for significantly higher doses which is associated with some adverse effects like oral moniliasis, vaginal moniliasis, hypertension, dyspepsia, localized abdominal pain, pruritis and tongue discoloration, myelosupression (including anemia, leukopenia, pancytopenia and thrombocytopenia), peripheral neuropathy and optic neuropathy sometimes progressing loss of vision and lactic acidosis. Linezolid has MIC of 1-4 μg/ml.
  • U.S. Pat. No. 6,514,529 (“the '529 patent”) discloses immediate release tablets containing a high drug content of oxazolidinones such as Linezolid, wherein the oxazolidinone is compressed into a tablet using corn starch, microcrystalline cellulose, hydroxylpropyl cellulose, sodium starch glycolate and magnesium stearate as the excipients. The '529 patent further discloses method for providing blood levels of Linezolid by oral administration medically equivalent to the blood levels produced by IV administration of the Linezolid which comprises administration of immediate release tablet formulation.
  • U.S. Pat. No. 6,451,345 disclose taste masked microcapsule compositions for oxazolidinone or macrolide antibiotics comprise microcapsules of drug coated by coacervation of a microencapsulation polymer and a coated plasticized enteric polymer.
  • However, no reference till now discloses any extended release compositions of Linezolid suitable for once daily dosing.
  • Extended or sustained or slow release compositions offer clinically significant advantages for various therapeutically active agents by way of increasing patient compliance due to reduced frequency of administration; improve the safety and efficacy of drug substances and reduce undesirable effects in comparison to the corresponding immediate release dosage form.
  • When immediate-release compositions are given higher peak plasma concentrations are achieved this sometimes may be undesirable due to toxic effects. As extended release formulations releases the drug over prolonged period of time in controlled release manner the plasma concentrations may be achieved and maintained which are below toxic levels. Immediate release compositions in case of antibiotics due to the shorter half life eliminates faster from body and the plasma concentration drop down below MIC level and may not be effective; therefore for immediate release formulations the frequency of dosing required is more. In case of extended release formulations due to slow release of drug the in-vivo plasma concentrations are maintained above MIC level for longer period of time therefore reducing dosing frequency and improving efficacy which is desirable especially for antibiotics.
  • Linezolid belongs to antibiotics or antibacterial category which requires high dose of drug frequently (BID) to achieve required MIC level. For antibiotics, it is recognized that the time above minimum inhibitory concentration (T>MIC) is the pharmacodynamic parameter most closely related to efficacy and insufficient T>MIC may lead to bacterial resistance. A further parameter which may be of importance is the ratio of the maximum plasma concentration (Cmax) to the MIC value, as this may be related to the potential to select for resistance by microorganism.
  • Linezolid is high dose (1200/day) and high solubility (3 mg/ml) active. These types of actives pose challenges to a formulator as it is difficult to formulate an extended release dosage form which can be suitable for once daily dosing for such actives because:
      • The dose of active will be high and hence there will be very little scope to include release controlling materials in the dosage form.
      • The large quantities of release controlling materials are required to achieve controlled drug release over the period of time and which can be suitable for once daily dosing.
      • Use of large quantities release controlling materials causes increase in the size of the dosage form which will make it difficult to swallow for patients.
  • Thus there exists an unmet need to provide a stable, simple, cost effective extended release composition comprising Linezolid without unwanted adverse effects and which maintains therapeutic plasma levels above MIC for a longer period of time.
  • SUMMARY OF THE INVENTION
  • It is, therefore, an object of the present invention to provide an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • In yet another aspect of the present invention provides an extended release solid pharmaceutical composition suitable for once daily dosing such as tablet comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • A further aspect of the present invention provides a process for the preparation of an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • BRIEF DESCRIPTION OF THE DRAWING
  • FIG. 1 shows dissolution profiles of the pharmaceutical compositions according to the present invention for Example 1 to Example 19 using USP Type II (Paddle Apparatus) at 50 rpm in 900 ml/1000 ml, 0.01N HCl for first 2 hours, followed by the media with pH 6.8 Phosphate buffer.
  • FIG. 2 shows comparative dissolution profiles of the pharmaceutical compositions according to the present invention for Example 11 and Example 15 using USP Type II (Paddle Apparatus) at 50 rpm in 900 ml/1000 ml, 0.1N HCl, pH 4.5 Acetate buffer and pH 6.8 Phosphate buffer.
  • FIG. 3 shows comparative mean plasma concentration (μg/ml) vs. time (hours) profile for 1200 mg (600 mg×2 Tablets), 1000 mg ER tablets according to Example 11 (Test 1) and Example 15 (Test 2) respectively and reference immediate release Zyvox 600 mg tablet twice daily.
  • DETAILED DESCRIPTION OF THE INVENTION
  • In describing and claiming the present invention, the following terminology will be used in accordance with the definitions set forth below.
  • The term “Linezolid” herein refers to any pharmaceutically acceptable form of Linezolid including base or its pharmaceutically acceptable complexes, salts, prodrug, derivative polymorphs, hydrates, solvates, enantiomers or racemates, metabolite as dictated by the context of its use. Linezolid may be present from about 50 mg to about 1700 mg Linezolid; preferably from about 300 mg to about 1500 mg Linezolid, preferably about 600 mg, about 1000 mg, about 1200 mg or about 1500 mg of Linezolid.
  • The term “extended release pharmaceutical composition” herein refers to any composition or dosage form that comprises an active drug and which is formulated to provide a longer duration of pharmacological response after administration of the dosage form than is ordinarily experienced after administration of a corresponding immediate release composition. Extended release compositions include, inter alia, those compositions described elsewhere as “controlled release”, “delayed release”, “sustained release”, “prolonged release”, “programmed release”, “time release” and/or “rate controlled” compositions or dosage forms.
  • The term “pharmaceutically acceptable” is meant a carrier comprised of a material that is not biologically or otherwise undesirable.
  • “Cmax” as used herein, means maximum plasma concentration of Linezolid, produced by the oral administration of the composition of the invention or the immediate release (IR) comparator.
  • “Tmax” as used herein, means time to the maximum observed plasma concentration.
  • “AUC0-24” as used herein, means area under the plasma concentration-time curve, as calculated by the trapezoidal rule over the complete 24-hour interval for all the formulations.
  • “T>MIC” as used herein, means time above MIC (minimum inhibitory concentration) calculated manually by graphical interpolation, where the minimum inhibitory plasma concentration was defined as 2 μg/ml of Linezolid.
  • “Adverse effects” as used herein, means those physiological effects to various systems in the body such as cardiovascular systems, nervous system, digestive system, and body as a whole, which cause pain and discomfort to the individual subject.
  • The various embodiments of the present invention can be assembled in several different ways.
  • In one embodiment the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • In another embodiment the present invention provides an extended release tablet suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • In yet another embodiment the present invention provides an extended release matrix tablet suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • In yet another embodiment the present invention provides an extended release coated tablet suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • In yet another embodiment the present invention provides a process of preparing an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
  • In yet another embodiment the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more release controlling material(s).
  • In yet another embodiment the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more hydrophilic release controlling material(s).
  • In yet another embodiment the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more hydrophobic release controlling material(s).
  • In yet another embodiment the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and a matrix comprising one or more hydrophilic release controlling material(s) optionally coated with hydrophilic material and/or hydrophobic material or combinations thereof.
  • In yet another embodiment the present invention provides an extended release tablet suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more hydrophilic release controlling material(s) optionally coated with hydrophilic material and/or hydrophobic material or combinations thereof.
  • In yet another embodiment the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and pharmaceutically acceptable excipient having a in-vitro dissolution rate when measured using the USP Type II (Paddle apparatus) at 50 rpm in 900 ml/1000 ml, 0.01N hydrochloric acid for first 2 hours followed by the media with pH 6.8 phosphate buffer at 37° C. from about 5 to about 50% Linezolid released after 1 hour; from about 10 to about 95% Linezolid released after 4 hours; from about 35 to about 100% Linezolid released after 8 hours; from about 55 to about 100% Linezolid released after 12 hours; from about 70 to about 100% Linezolid released after 16 hours; and greater than 90% Linezolid released after 24 hours.
  • In yet another embodiment the present invention provides an extended release tablet comprising Linezolid having two layers comprising immediate release layer and controlled release layer; wherein immediate release layer comprises of loading dose and controlled release layer comprises of maintenance dose wherein loading dose is released immediately and maintenance dose released in controlled manner over a period of time.
  • In yet another embodiment the present invention provides an extended release tablet suitable for once daily dosing comprising:
    • a) 50-1500 mg of Linezolid or pharmaceutically acceptable salt, derivative, prodrug, and metabolite thereof;
    • b) 0.1% to 20% w/w of one or more release controlling materials;
    • c) one or more other pharmaceutically acceptable excipients.
  • In yet another embodiment the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients so that upon oral administration the maximum concentrations (Cmax) of Linezolid in plasma are statistically significantly lower than the immediate release formulation given twice daily, and area under the plasma concentration-time curve (AUC) and the minimum plasma concentration are maintained over 24 hours.
  • In yet another embodiment the present invention provides an extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients so that upon oral administration, the composition induces lower fluctuation in the mean plasma concentration than an immediate release composition of the Linezolid.
  • In yet another embodiment the present invention provides a method of using an extended release, pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof as an active ingredient, and one or more pharmaceutically acceptable excipients, comprising administering the composition in an effective amount for the treatment for bacterial infection in a mammal.
  • In yet another embodiment the present invention provides a method of treating bacterial infections in a mammal comprising administering an extended release, pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof as an active ingredient, and one or more pharmaceutically acceptable excipients, capable of maintaining a serum level of the Linezolid above MIC level (2 μg/ml) for at least 24 hours.
  • In yet another embodiment the present invention provides method of treating bacterial infections in a mammal comprising administering an extended release, pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof as an active ingredient, and one or more pharmaceutically acceptable excipients, capable of maintaining T>MIC for at least 24 hours.
  • In a preferred embodiment an extended release pharmaceutical composition according to present invention comprises of Linezolid from about 10% w/w to about 95% w/w of the composition, preferably from about 50% w/w to about 95% w/w of the composition.
  • In a preferred embodiment an extended release pharmaceutical compositions of present invention upon oral administration to a human, provides a mean maximum plasma concentration (Cmax) of Linezolid not more than, about 16 μg/ml.
  • The active ingredient in the preparation according to the invention may suitably be incorporated in a matrix. This may be any matrix that affords extended release of Linezolid that affords in-vitro dissolution rates over 24 hours. Preferably the matrix is an extended release matrix. Alternatively, immediate release matrices having a release controlling coating or controlled release matrices having immediate release coating, which provides extended release of the active ingredient, may be used.
  • Tableting is preferred production method because it is faster, easier, adds fewer steps to the process and is the most economical. Further, the tableting method ensures a high production yield, contrary to the manufacture of pellets where the loss of production output is usually much higher. Excipients for the formulation were chosen carefully to give appropriate dissolution rate and stability of the finished dosage form.
  • In yet another embodiment the present invention provides, a process for preparing an extended release pharmaceutical composition comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, and metabolite thereof and water soluble binder wherein process can be selected from direct compression, dry granulation, wet granulation (aqueous/non-aqueous or combination) and melt granulation.
  • In a preferred embodiment an extended release composition according to present invention comprises of one or more release controlling materials. The release controlling materials can be hydrophilic release controlling materials or hydrophobic release controlling materials. The release controlling materials are present from about 0.1% to about 20% w/w of the composition.
  • Examples of suitable hydrophilic release controlling materials according to present invention include but are not limited to hydroxypropyl methylcellulose, hydroxypropyl cellulose, methylcellulose, povidone, polyethylene glycols, vinyl acetate copolymers, polysaccharides such as alginates, xanthan gum, chitosan, carrageenan, dextran and the like, polyalkylene oxides such as polyethylene oxide and the likes, methacrylic acid copolymers, maleic anhydride/methyl vinyl ether copolymers, carbomers and the like.
  • Examples of hydrophobic release controlling materials according to present invention include but are not limited to polyvinyl acetate dispersion, ethyl cellulose, cellulose acetate, cellulose propionate (lower, medium or higher molecular weight), cellulose acetate propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose triacetate, poly (methyl methacrylate), poly (ethyl methacrylate), poly (butyl methacrylate), poly (isobutyl methacrylate), and poly (hexyl methacrylate), poly (isodecyl methacrylate), poly (lauryl methacrylate), poly (phenyl methacrylate), poly (methyl acrylate), poly (isopropyl acrylate), poly (isobutyl acrylate), poly (octadecyl acrylate), waxes such as beeswax, carnauba wax, paraffin wax, microcrystalline wax, ozokerite; fatty alcohols such as cetostearyl alcohol, stearyl alcohol, cetyl alcohol, myristyl alcohol, and fatty acid esters such as glyceryl monostearate, glycerol monooleate, acetylated monoglycerides, tristearin, tripalmitin, cetyl esters wax, glyceryl palmitostearate, glyceryl behenate, zein and hydrogenated vegetable oils or their mixtures thereof.
  • The formulation will, in general comprise of one or more excipients. Examples of pharmaceutically acceptable excipients include, but are not limited to, diluents, disintegrants, lubricant, glidant, binders, fillers, surfactant, solubilizers, wetting agents, chelating agents, stabilizers, alkalizing agents or amino acids. A combination of excipients may also be used. The amount of excipient(s) employed will depend upon how much active agent is to be used. One excipient can perform more than one function.
  • Binders include, but are not limited to, starches such as potato starch, wheat starch, corn starch; microcrystalline cellulose such as products known under the registered trade marks Avicel, Filtrak, Heweten or Pharmacel; celluloses such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxypropylmethyl cellulose (HPMC), ethyl cellulose, sodium carboxy methyl cellulose; natural gums like acacia, alginic acid, guar gum; liquid glucose, dextrin, povidone, syrup, polyethylene oxide, polyvinyl pyrrolidone, poly-N-vinyl amide, polyethylene glycol, gelatin, poly propylene glycol, tragacanth, combinations there of and other materials known to one of ordinary skill in the art.
  • Fillers or diluents, which include, but are not limited to confectioner's sugar, compressible sugar, dextrates, dextrin, dextrose, fructose, lactitol, mannitol, sucrose, starch, lactose, xylitol, sorbitol, talc, microcrystalline cellulose, calcium carbonate, calcium phosphate dibasic or tribasic, calcium sulphate, and the like can be used.
  • Lubricants may be selected from, but are not limited to, those conventionally known in the art such as Mg, Al, Ca or Zn stearate, polyethylene glycol, glyceryl behenate, mineral oil, sodium stearyl fumarate, stearic acid, hydrogenated vegetable oil and talc.
  • Glidants include, but are not limited to, silicon dioxide, magnesium trisilicate, powdered cellulose, starch, talc and tribasic calcium phosphate, calcium silicate, magnesium silicate, colloidal silicon dioxide, silicon hydrogel and other materials known to one of ordinary skill in the art.
  • The pharmaceutical composition according to the present invention include but is not limited to tablets (single layered tablets, multilayered tablets, mini tablets, bioadhesive tablets, caplets, matrix tablets, tablet within a tablet, mucoadhesive tablets, modified release tablets, pulsatile release tablets, timed release tablets), pellets, beads, granules, sustained release formulations, capsules, microcapsules, tablets in capsules and microspheres, matrix formulations, microencapsulation and powder/pellets/granules for suspension.
  • The pharmaceutical composition of the invention can optionally have one or more coatings such as film coating, sugar coating, enteric coating, bioadhesive coating and other coatings known in the art. These coatings help pharmaceutical formulations to release the drug at the required site of action.
  • In one example, the additional coating prevents the dosage form from contacting the mouth or esophagus. In another example, the additional coating remains intact until reaching the small intestine (e.g., an enteric coating). Premature exposure of a bioadhesive layer or dissolution of a pharmaceutical dosage form in the mouth can be prevented with a layer or coating of hydrophilic polymers such as HPMC or gelatin. Optionally, Eudragit FS 30D or other suitable polymer may be incorporated in coating composition to retard the release of the drug to ensure drug release in the colon.
  • These coating layers comprises one or more excipients selected from the group comprising coating agents, opacifiers, taste-masking agents, fillers, polishing agents, colouring agents, antitacking agents and the like.
  • Coating agents which are useful in the coating process, include, but are not limited to, polysaccharides such as maltodextrin, alkyl celluloses such as methyl or ethyl cellulose, hydroxyalkylcelluloses (e.g. hydroxypropylcellulose or hydroxypropylmethylcelluloses); polyvinylpyrrolidone, acacia, corn, sucrose, gelatin, shellac, cellulose acetate pthalate, lipids, synthetic resins, acrylic polymers, opadry, polyvinyl alcohol (PVA), copolymers of vinylpyrrolidone and vinyl acetate (e.g. marketed under the brand name of Plasdone) and polymers based on methacrylic acid such as those marketed under the brand name of Eudragit. These may be applied from aqueous or non-aqueous systems or combinations of aqueous and non-aqueous systems as appropriate. Additives can be included along with the film formers to obtain satisfactory films. These additives can include plasticizers such as dibutyl phthalate, triethyl citrate, polyethylene glycol (PEG) and the like, antitacking agents such as talc, stearic acid, magnesium stearate and colloidal silicon dioxide and the like, surfactants such as polysorbates and sodium lauryl sulphate, fillers such as talc, precipitated calcium carbonate, polishing agents such as beeswax, carnauba wax, synthetic chlorinated wax and opacifying agents such as titanium dioxide and the like. All these excipients can be used at levels well known to the persons skilled in the art.
  • Pharmaceutical composition of the invention can be coated by a wide variety of methods. Suitable methods include compression coating, coating in a fluidized bed or a pan and hot melt (extrusion) coating. Such methods are well known to those skilled in the art.
  • Non-permeable coatings of insoluble polymers, e.g., cellulose acetate, ethylcellulose, can be used as enteric coatings for delayed/modified release (DR/MR) by inclusion of soluble pore formers in the coating, e.g., PEG, PVA, sugars, salts, detergents, triethyl citrate, triacetin, etc.
  • Multi-layer or gradient tablets can be assembled in several different ways.
  • The invention will be further illustrated by the following examples, which are intended to be illustrative but not limiting.
  • Example No. 1
  • Ingredients Weight (mg)
    Linezolid 600.0
    Microcrystalline Cellulose 176.2
    Polyacrylate dispersion 20.0
    30 percent
    (Eudragit NE 30D)
    Purified water q.s.
    Magnesium Stearate 3.8
    Total 800.0
  • Manufacturing Procedure:
  • Linezolid and microcrystalline cellulose were sifted through suitable sieve and mixed. The blend was granulated with Eudragit NE 30D and sufficient quantity of purified water. The granules were dried and sifted through suitable sieve. The Magnesium Stearate was sifted through suitable sieve and mixed with granules. The blend was compressed into tablets.
  • Example No. 2
  • Ingredients Weight (mg)
    Linezolid 600.0
    Lactose Monohydrate 180.2
    Povidone 16.0
    Purified water q.s.
    Magnesium Stearate 3.8
    Total 800.0
  • Manufacturing Procedure:
  • Linezolid, lactose monohydrate and povidone were sifted through suitable sieve and mixed. The blend was granulated with sufficient quantity of purified water. The granules were dried and sifted through suitable sieve. The Magnesium Stearate was sifted through suitable sieve and mixed with granules. The blend was compressed into tablets.
  • Example No. 3
  • Ingredients Weight (mg)
    Linezolid 1200.0
    Lactose Monohydrate 356.4
    Povidone 16.0
    Purified water q.s.
    Magnesium Stearate 7.6
    Total 1580.0
  • Manufacturing Procedure:
  • Linezolid, lactose monohydrate and povidone were sifted through suitable sieve and mixed. The blend was granulated with sufficient quantity of purified water. The granules were dried and sifted through suitable sieve. The Magnesium Stearate was sifted through suitable sieve and mixed with granules. The blend was compressed into tablets.
  • Example No. 4
  • Ingredients Weight (mg)
    Linezolid 1000.0
    Lactose Monohydrate 297.0
    Povidone 13.3
    Purified water q.s.
    Magnesium Stearate 6.3
    Total 1316.6
  • Manufacturing Procedure:
  • Linezolid, lactose monohydrate and povidone were sifted through suitable sieve and mixed. The blend was granulated with sufficient quantity of purified water. The granules were dried and sifted through suitable sieve. The Magnesium Stearate was sifted through suitable sieve and mixed with granules. The blend was compressed into tablets.
  • Example No. 5
  • Ingredients Weight (mg)
    Linezolid 600.0
    Lactose Monohydrate 290.0
    Povidone 8.0
    Purified water q.s.
    Magnesium Stearate 2.0
    Total 900.0
  • Manufacturing Procedure:
  • Linezolid, lactose monohydrate and povidone were sifted through suitable sieve and mixed. The blend was granulated with sufficient quantity of purified water. The granules were dried and sifted through suitable sieve. The Magnesium Stearate was sifted through suitable sieve and mixed with granules. The blend was compressed into tablets.
  • Example No. 6
  • Ingredients Weight (mg)
    Linezolid 600.0
    Lactose Monohydrate 25.0
    Polyethylene Glycol 6000 50.0
    Povidone 20.0
    Purified water q.s.
    Magnesium Stearate 5.0
    Total 700.0
  • Manufacturing Procedure:
  • Linezolid, Polyethylene Glycol 6000, lactose monohydrate and povidone were sifted through suitable sieve and mixed. The blend was granulated with sufficient quantity of purified water. The granules were dried and sifted through suitable sieve. The Magnesium Stearate was sifted through suitable sieve and mixed with granules. The blend was compressed into tablets.
  • Example No. 7
  • Ingredients Weight (mg)
    Linezolid 600.0
    Lactose Monohydrate 90.0
    Ethyl Cellulose 35.0
    Povidone 20.0
    Purified water q.s.
    Magnesium Stearate 5.0
    Total 750.0
  • Manufacturing Procedure:
  • Linezolid, lactose monohydrate and ethylcellulose were sifted through suitable sieve and mixed. The povidone was dissolved in sufficient quantity of purified water. The blend was granulated using povidone solution. The granules were dried and sifted through suitable sieve. The Magnesium Stearate was sifted through suitable sieve and mixed with granules. The blend was compressed into tablets.
  • Example No. 8
  • Ingredients Weight (mg)
    Linezolid 600.0
    Lactose Monohydrate 250.0
    Hydroxypropyl Cellulose 20.0
    Purified water q.s.
    Talc 15.0
    Magnesium Stearate 15.0
    Total 900.0
  • Manufacturing Procedure:
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried and sifted. Talc and magnesium stearate were sifted and mixed with granules. The blend was compressed into tablets.
  • Example No. 9 Bi-Layer Composition
  • Ingredients Weight (mg)
    Immediate Release Layer
    Linezolid 150.0
    Corn Starch 15.0
    Microcrystalline Cellulose 6.0
    Hydroxypropyl cellulose 3.0
    Purified Water q.s.
    Microcrystalline Cellulose 35.9
    Sodium Starch Glycolate 10.5
    Talc 2.35
    Magnesium Stearate 2.25
    Total 225.0
    Controlled Release Layer
    Linezolid 450.0
    Lactose monohydrate 180.0
    Povidone 22.5
    Purified Water q.s.
    Talc 11.25
    Magnesium Stearate 11.25
    Total 675.0
    Total Weight of Bilayer 900.0
    Tablet
  • Manufacturing Procedure: Immediate Release Layer
  • Linezolid, corn starch, microcrystalline cellulose and hydroxypropylcellulose were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate. The blend was mixed with sifted talc and magnesium stearate.
  • Controlled Release Layer
  • Linezolid, lactose monohydrate and povidone were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried and sifted. The magnesium stearate was sifted and mixed with granules.
  • The blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablets.
  • Example No. 10
  • Ingredients Weight (mg)
    Linezolid 600.0
    Lactose Monohydrate 260.0
    Hydroxypropyl Cellulose 20.0
    Purified Water q.s.
    Talc 10.0
    Magnesium Stearate 10.0
    Total 900.0
  • Manufacturing Procedure:
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried and sifted. Talc and magnesium stearate was sifted and mixed with granules. The blend was compressed into tablets.
  • Example No. 11 Bi-Layer Composition
  • Ingredients Weight (mg)
    Strength 600 mg
    Immediate Release Layer
    Linezolid 150.0
    Microcrystalline Cellulose 6.0
    Hypromellose 3.0
    Carboxymethylcellulose 15.0
    Calcium
    Purified Water q.s.
    Microcrystalline Cellulose 36.0
    Sodium Starch Glycolate 10.5
    Talc 2.25
    Magnesium Stearate 2.25
    Total 225.0
    Controlled Release Layer
    Linezolid 450.0
    Lactose monohydrate 180.0
    Povidone 22.5
    Purified Water q.s.
    Talc 11.25
    Magnesium Stearate 11.25
    Total 675.0
    Total Weight of Bilayer 900.0
    Tablet
  • Manufacturing Procedure: Immediate Release Layer
  • Linezolid, microcrystalline cellulose, hypromellose and Carboxymethylcellulose Calcium were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate. The blend was mixed with sifted talc and magnesium stearate.
  • Controlled Release Layer
  • Linezolid, lactose monohydrate and povidone were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried and sifted. The granules were mixed with sifted talc and magnesium stearate.
  • The blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablet.
  • Example No. 12 Bi-Layer Composition
  • Ingredients Weight (mg)
    Strength 1000 mg
    Immediate Release Layer
    Linezolid 250.0
    Microcrystalline Cellulose 10.0
    Hypromellose 5.0
    Carboxymethylcellulose 25.0
    Calcium
    Purified Water q.s.
    Microcrystalline Cellulose 60.0
    Sodium Starch Glycolate 17.5
    Talc 3.75
    Magnesium Stearate 3.75
    Total 375.0
    Controlled Release Layer
    Linezolid 750.0
    Lactose monohydrate 300.0
    Povidone 37.5
    Purified Water q.s.
    Talc 18.75
    Magnesium Stearate 18.75
    Total 1125.0
    Total Weight of Bilayer 1500.0
    Tablet
  • Manufacturing Procedure: Immediate Release Layer
  • Linezolid, microcrystalline cellulose, hypromellose and Carboxymethylcellulose Calcium were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate. The blend was mixed with sifted talc and magnesium stearate.
  • Controlled Release Layer
  • Linezolid, lactose monohydrate and povidone were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried and sifted. The granules were mixed with sifted talc and magnesium stearate.
  • The blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablet.
  • Example No. 13 Compression Coated Tablet (Tablet in Tablet) Composition
  • Ingredients Weight (mg)
    Controlled Release Inner Core
    Linezolid 450.0
    Lactose monohydrate 73.0
    Povidone 15.0
    Purified Water q.s.
    Talc 6.0
    Magnesium Stearate 6.0
    Total Controlled Release Core 550.0
    Immediate Release outer compression Coat
    Linezolid 150.0
    Microcrystalline Cellulose 500.0
    Carboxymethylcellulose 60.0
    Calcium
    Hypromellose 15.0
    Purified Water q.s.
    Microcrystalline Cellulose 210.0
    Sodium Starch Glycolate 45.0
    Talc 10.0
    Magnesium Stearate 10.0
    Total 1000.0
    Total weight of compression 1550.0
    coated tablet
  • Manufacturing Procedure: Controlled Release Inner Core
  • Linezolid, lactose monohydrate and povidone were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried and sifted. The granules were mixed with sifted talc and magnesium stearate. The blend was compressed into tablet.
  • Immediate Release Outer Compression Coat
  • Linezolid, microcrystalline cellulose, hypromellose and Carboxymethylcellulose Calcium were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules was dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate. The blend was mixed with sifted talc and magnesium stearate. The inner core tablets were compression coated with the immediate release blend to form a controlled release tablet inside the immediate release tablets.
  • Example No. 14 Bi-Layer Composition
  • Ingredients Weight (mg)
    Strength 1000 mg
    Immediate Release Layer
    Linezolid 250.0
    Microcrystalline Cellulose 10.0
    Hypromellose 5.0
    Carboxymethylcellulose 25.0
    Calcium
    Purified Water q.s.
    Microcrystalline Cellulose 60.0
    Sodium Starch Glycolate 17.5
    Talc 3.75
    Magnesium Stearate 3.75
    Total 375.0
    Controlled Release Layer
    Linezolid 750.0
    Lactose monohydrate 312.5
    Hydroxypropyl Cellulose 25.0
    Purified Water q.s.
    Talc 18.75
    Magnesium Stearate 18.75
    Total 1125.0
    Total Weight of Bilayer 1500.0
    Tablet
  • Manufacturing Procedure: Immediate Release Layer
  • Linezolid, microcrystalline cellulose, hypromellose and Carboxymethylcellulose Calcium were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate. The blend was mixed with sifted talc and magnesium stearate.
  • Controlled Release Layer
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried and sifted. The granules were mixed with sifted talc and magnesium stearate.
  • The blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablet.
  • Example No. 15 Bi-Layer Composition
  • Ingredients Weight (mg)
    Strength 1000 mg
    Immediate Release Layer
    Linezolid 250.0
    Microcrystalline Cellulose 10.0
    Hypromellose 5.0
    Carboxymethylcellulose 25.0
    Calcium
    Purified Water q.s.
    Microcrystalline Cellulose 60.0
    Sodium Starch Glycolate 17.5
    Talc 3.75
    Magnesium Stearate 3.75
    Total 375.0
    Controlled Release Layer
    Linezolid 750.0
    Lactose monohydrate 105.0
    Povidone 25.0
    Purified Water q.s.
    Talc 10.0
    Magnesium Stearate 10.0
    Total 900.0
    Total Weight of Bilayer 1275.0
    Tablet
  • Manufacturing Procedure: Immediate Release Layer
  • Linezolid, microcrystalline cellulose, hypromellose and Carboxymethylcellulose Calcium were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried, sifted and mixed with sifted microcrystalline cellulose, sodium starch glycolate. The blend was mixed with sifted talc and magnesium stearate.
  • Controlled Release Layer
  • Linezolid, lactose monohydrate and povidone were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried and sifted. The granules were mixed with sifted talc and magnesium stearate.
  • The blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablet.
  • Example No. 16
  • Ingredients Weight (mg)
    Core Tablet
    Linezolid 600.0
    Microcrystalline Cellulose 190.0
    HPMC 200.0
    Purified Water q.s.
    Talc 10.0
    Magnesium Stearate 10.0
    Total 1010.0
  • Manufacturing Procedure:
  • Linezolid, microcrystalline cellulose and HPMC were sifted and mixed. The blend was granulated with purified water. The granules were dried, sifted and mixed with sifted talc and magnesium stearate. The blend was compressed into tablets.
  • Example 17 Bilayer Tablet
  • Ingredients Weight (mg)
    Strength 600 mg
    Immediate Release Layer
    Linezolid 150.0
    Dibasic Calcium Phosphate 48.0
    Croscarmellose Sodium 5.0
    Carboxymethylcellulose 15.0
    Calcium
    Purified Water q.s.
    Croscarmellose Sodium 5.0
    Magnesium Stearate 2.0
    Total 225.0
    Controlled Release Layer
    Linezolid 450.0
    Lactose monohydrate 180.0
    Hydroxypropylcellulose 22.5
    Purified Water q.s.
    Talc 11.25
    Magnesium Stearate 11.25
    Total 675.0
    Total Weight of Bilayer 900.0
    Tablet
  • Manufacturing Procedure:
  • Immediate Release Layer
  • Linezolid, dibasic calcium phosphate, croscarmellose sodium and carboxymethylcellulose calcium were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried, sifted and mixed with sifted croscarmellose sodium. The blend was mixed with sifted magnesium stearate.
  • Controlled Release Layer
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried and sifted. The granules were mixed with sifted talc and magnesium stearate.
  • The blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablets.
  • Example 18 Bilayer Tablet
  • Ingredients Weight (mg)
    Strength 1000 mg
    Immediate Release Layer
    Linezolid 250.0
    Dibasic Calcium Phosphate 80.0
    Croscarmellose Sodium 8.3
    Carboxymethylcellulose 25.0
    Calcium
    Purified Water q.s.
    Croscarmellose Sodium 8.3
    Magnesium Stearate 3.4
    Total 375.0
    Controlled Release Layer
    Linezolid 750.0
    Lactose monohydrate 300.0
    Hydroxypropylcellulose 37.5
    Purified Water q.s.
    Talc 18.75
    Magnesium Stearate 18.75
    Total 1125.0
    Total Weight of Bilayer 1500.0
    Tablet
  • Manufacturing Procedure: Immediate Release Layer
  • Linezolid, dibasic calcium phosphate, croscarmellose sodium and carboxymethylcellulose calcium was sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules was dried, sifted and mixed with sifted croscarmellose sodium. The blend was mixed with sifted magnesium stearate.
  • Controlled Release Layer
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried and sifted. The granules were mixed with sifted talc and magnesium stearate.
  • The blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablets.
  • Example 19 Bilayer Tablet
  • Ingredients Weight (mg)
    Strength 1000 mg
    Immediate Release Layer
    Linezolid 250.0
    Dibasic Calcium Phosphate 80.0
    Croscarmellose Sodium 8.3
    Carboxymethylcellulose 25.0
    Calcium
    Purified Water q.s.
    Croscarmellose Sodium 8.3
    Magnesium Stearate 3.4
    Total 375.0
    Controlled Release Layer
    Linezolid 750.0
    Lactose monohydrate 105.0
    Hydroxypropylcellulose 25.0
    Purified Water q.s.
    Talc 10.0
    Magnesium Stearate 10.0
    Total 900.0
    Total Weight of Bilayer 1275.0
    Tablet
  • Manufacturing Procedure: Immediate Release Layer
  • Linezolid, dibasic calcium phosphate, croscarmellose sodium and carboxymethylcellulose calcium were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried, sifted and mixed with sifted croscarmellose sodium. The blend was mixed with sifted magnesium stearate.
  • Controlled Release Layer
  • Linezolid, lactose monohydrate and hydroxypropylcellulose were sifted and mixed. The blend was granulated using sufficient quantity of purified water. The granules were dried and sifted. The granules were mixed with sifted talc and magnesium stearate.
  • The blend of immediate release layer was compressed over the pre-compressed blend of controlled release layer to form a bilayer tablets.
  • Example No. 20 Coated Tablet
  • Ingredients Weight (mg)
    Core Tablet
    Linezolid 600.0
    Corn Starch 60.0
    Microcrystalline Cellulose 24.0
    Hydroxypropylcellulose 12.0
    Purified Water q.s.
    Microcrystalline Cellulose 153.6
    Sodium Starch Glycolate 42.0
    Magnesium Stearate 8.4
    Total 900.0
    Controlled Release Coating
    Ethylcellulose based 45.0
    coating material
    (Surelease)
    HPMC based coating 6.75
    material (Opadry clear)
    Purified Water q.s.
    Total coating 51.75
  • Manufacturing Procedure: Core Tablet
  • Linezolid, corn starch, microcrystalline cellulose and hydroxypropylcellulose were sifted and mixed. The blend was granulated with sufficient quantity of purified water. The granules were dried, sifted and mixed with microcrystalline cellulose and sodium starch glycolate. The blend was mixed with magnesium stearate and compressed into tablets.
  • Coating: HPMC based coating material (Opadry Clear) was dissolved in sufficient quantity of purified water and mixed with ethylcellulose based aqueous coating material (Surelease). The core tablets were coated with the coating solution to a desired weight gain.
  • Example No. 21 Coated Tablet
  • Ingredients Weight (mg)
    Core Tablet
    Linezolid 600.0
    Corn Starch 60.0
    Microcrystalline Cellulose 24.0
    Hydroxypropylcellulose 12.0
    Purified Water q.s.
    Microcrystalline Cellulose 143.0
    Sodium Starch Glycolate 42.0
    Talc 10.0
    Magnesium Stearate 9.0
    Total 900.0
    Controlled Release Coating
    Ethylcellulose 45.0
    HPMC 18.0
    Triethylcitrate 4.5
    Isopropyl Alcohol q.s.
    Purified Water q.s.
    Total coating 67.5
  • Manufacturing Procedure: Core Tablet
  • Linezolid, corn starch, microcrystalline cellulose and hydroxypropylcellulose were sifted and mixed. The blend was granulated with sufficient quantity of purified water. The granules were dried, sifted and mixed with microcrystalline cellulose and sodium starch glycolate. The blend was mixed with magnesium stearate and compressed into tablets.
  • Coating: HPMC, triethylcitrate was dissolved in sufficient quantity of purified water, ethylcellulose was dissolved in isopropyl alcohol. Both the solutions were mixed and the core tablets were coated to a desired weight gain.
  • Example No. 22 Pharmacokinetic Study of the Extended Release Linezolid Formulation
  • The pharmacokinetic study to determine the concentration-time plasma profile was done on healthy male subjects. The study was conducted as open label, balanced, randomized, three-treatment, single-period, parallel, comparative oral bioavailability study as described below:
  • Single Dose Study:
  • A total of 18 normal, healthy, adult, male subjects were enrolled in the study. Healthy male subjects were aged between 18 and 40 years with BMI between 19-24 Kg/m2 but body weight not less than 45 Kgs.
  • The Linezolid ER tablets 1200 mg (600 mg×2 tablets as a single dose) and Linezolid ER tablets 1000 mg (as a single dose) and Linezolid 600 mg IR Tablets (reference product) currently sold by Pharmacia Limited under the Trade name Zyvox® (2 times at the interval of 12 hours (2 doses)) were administered to 18 healthy subjects under fasting condition in the morning.
  • The study was conducted according to open label, balanced, randomized, three-treatment, single-period, parallel, comparative oral bioavailability study.
  • The blood samples (5 ml each) were withdrawn at pre-dose (before dosing, in the morning of the day of dosing) and at 0.5, 1.0, 2.0, 3.0, 4.0, 6.0, 8.0, 12.0, 16.0, 24.0, 36.0 and 40.0 hours after morning dosing. Plasma samples were analyzed to quantify the concentrations of Linezolid using a validated LC/MS/MS bioanalytical method.
  • Pharmacokinetic Analyses:
  • Values for Linezolid pharmacokinetic parameters including observed AUC0-24 Cmax, Tmax, were calculated using standard non-compartmental methods. The time above the minimum inhibitory plasma concentration (T>MIC) was calculated manually by graphical interpolation, where the minimum inhibitory plasma concentration was defined as 2 μg/ml of Linezolid.
  • The in vitro dissolution is carried out using USP Type II (Paddle Apparatus) at 50 rpm in 900 ml/1000 ml, 0.01N HCl first two hours, followed by the media with pH 6.8 Phosphate buffer. The release of drug is measured using UV techniques. The dissolution pattern for Example No. 1 to Example No. 9 is given in the table below
  • Dissolution
    Dissolution Media: 900 ml/1000 ml 0.01N HCl for first 2 hours
    followed by media pH 6.8 Phosphate Buffer, USP II, 50 RPM
    Example No.
    1 2 3 4 5 6 7 8 9
    Time (Hours) Cumulative % Drug Released
    1 41 10 9 9 13 11 9 10 33
    2 67 18 17 17 25 19 13 18 39
    4 96 32 29 28 42 33 22 37 48
    6 100 40 38 37 55 43 30 54 54
    8 49 46 45 66 54 38 83 59
    10 58 53 52 76 62 45 85 64
    12 66 59 58 84 69 53 107 69
    14 71 65 64 92 76 59 74
    16 79 70 70 99 82 67 79
    20 91 80 80 93 78 87
    24 101 89 89 101 88 92
  • The in vitro dissolution is carried out using USP Type II (Paddle Apparatus) at 50 rpm in 900 ml/1000 ml, 0.01N HCl first two hours, followed by the media with pH 6.8 Phosphate buffer. The release of drug is measured using UV techniques. The dissolution pattern for Example No. 10 to Example No. 19 is given in the table below:
  • Dissolution
    Dissolution Media: 900 ml/1000 ml, 0.01N HCl for first 2 hours
    followed by media pH 6.8 Phosphate Buffer, USP II, 50 RPM
    Example
    10 11 12 13 14 15 16 17 18 19
    Time (hours) Cumulative % Drug Released
    1 43 33 30 32 30 30 6 35 34 33
    2 68 39 35 34 37 36 11 42 40 41
    4 91 49 43 45 51 45 22 59 50 51
    6 94 55 49 53 66 52 27 75 60 60
    8 105 60 54 61 82 58 33 89 71 70
    10 66 59 67 93 63 41 98 81 78
    12 71 63 73 98 68 50 101 88 86
    14 75 66 78 100 71 57 95 92
    16 80 69 83 76 64 100 98
    20 88 75 91 80 73
    24 95 80 97 84 85
  • The in vitro dissolution is carried out using USP Type II (Paddle Apparatus) at 50 rpm in 900 ml/1000 ml, 0.1 N HCl, pH 4.5 Acetate Buffer and pH 6.8 Phosphate Buffer. The release of drug is measured using UV techniques. The dissolution pattern for Example No. 11 and Example No. 15 is given in the table below:
  • Strength
    600 mg 1000 mg 600 mg 1000 mg 600 mg 1000 mg
    Example No.
    11 15 11 15 11 15
    Media 0.1N HCl pH 4.5 Acetate Buffer pH 6.8 Phosphate Buffer
    Volume 900 ml 1000 ml 900 ml 1000 ml 900 ml 1000 ml
    Condition USP II (Paddle), 50 RPM
    Time (Hrs) Cumulative % Drug Release
    0.5 35 33 27 27 30 27
    1 43 41 31 29 32 29
    2 54 52 35 33 36 33
    4 68 69 43 40 44 40
    6 76 74 50 45 50 45
    8 81 80 56 51 57 51
    10 86 83 60 56 61 55
    12 89 86 64 60 67 61
    14 92 87 66 64 71 65
    16 94 89 69 67 76 67
    20 97 93 74 72 86 77
    24 98 97 78 79 95 85
  • Values for Linezolid pharmacokinetic parameters as per pharmacokinetic study conducted in Example 22 including observed Cmax, Tmax, AUC0-24 T>MIC are given in below table.
  • Tmax (hrs) AUC0-24
    Cmax (μg/ml) Median (μg · h/ml) T > MIC
    Formulation Mean ± SD (Range) Mean ± SD (hrs)
    Example 11 Linezolid ER Tablet 1200 mg 8.16 ± 0.69 2.9(1.5-4.0) 119.70 ± 9.58  24
    (i.e. 600 mg × 2 tablets) as single dose.
    Example 15 Linezolid ER Tablet (1000 mg) 7.84 ± 1.06 2.0(0.5-3.5) 117.60 ± 38.00 24
    single dose.
    Reference Zyvox ® (600 mg) 2 times at the 18.78 ± 3.58  1.5(0.5-2.5) 334.72 ± 45.61 24
    interval of 12 hours (2 doses)

Claims (44)

1. An extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients.
2. An extended release pharmaceutical composition according to claim 1 wherein the Linezolid is present from about 50 mg to about 1700 mg.
3. An extended release pharmaceutical composition according to claim 1 wherein the Linezolid is present from about 300 mg to about 1500 mg.
4. An extended release pharmaceutical composition according to claim 1 wherein the Linezolid is present from about 10% w/w to about 95% w/w of the composition.
5. An extended release pharmaceutical composition according to claim 1 wherein the Linezolid is present from about 50% w/w to about 95% w/w of the composition.
6. An extended release pharmaceutical composition according to claim 1 which upon oral administration to a human provides a mean maximum plasma concentration (Cmax) of Linezolid not more than about 16 μg/ml.
7. An extended release pharmaceutical composition according to claim 1 which upon oral administration to a human provides a mean plasma concentration of at least 2 μg/ml of Linezolid for at least 24 hours.
8. A method of reducing adverse effects comprising administering pharmaceutical composition according to claim 1.
9. An extended release pharmaceutical composition according to claim 1 which upon oral administration induces lower fluctuation in the mean plasma concentration than an immediate release composition of the Linezolid.
10. An extended release pharmaceutical composition according to claim 1 wherein pharmaceutical composition comprises of one or more release controlling materials.
11. An extended release pharmaceutical composition according to claim 10 wherein release controlling materials are present from about 0.1% w/w to about 20% w/w of the composition.
12. An extended release pharmaceutical composition according to claim 10 wherein the release controlling material is selected from group consisting of hydrophilic release controlling material or hydrophobic release controlling material or combinations thereof.
13. An extended release pharmaceutical composition according to claim 12 wherein hydrophilic release controlling material is selected from group consisting of hydroxypropyl methylcellulose, hydroxypropyl cellulose, methylcellulose, povidone, polyethylene glycols, vinyl acetate copolymers, polysaccharides as alginates, xanthan gum, chitosan, carrageenan, dextran, polyalkylene oxides as polyethylene oxide, methaacrylic acid copolymers, maleic anhydride/methyl vinyl ether copolymers, carbomers.
14. An extended release pharmaceutical composition according to claim 12 wherein hydrophobic release controlling material is selected from group consisting of polyvinyl acetate dispersion, ethyl cellulose, cellulose acetate, cellulose propionate (lower, medium or higher molecular weight), cellulose acetate propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose triacetate, poly (methyl methacrylate), poly (ethyl methacrylate), poly (butyl methacrylate), poly (isobutyl methacrylate), poly (hexyl methacrylate), poly (isodecyl methacrylate), poly (lauryl methacrylate), poly (phenyl methacrylate), poly (methyl acrylate), poly (isopropyl acrylate), poly (isobutyl acrylate), poly (octadecyl acrylate), beeswax, carnauba wax, paraffin wax, microcrystalline wax, ozokerite; cetostearyl alcohol, stearyl alcohol, cetyl alcohol; myristyl alcohol, glyceryl monostearate; glycerol monooleate, acetylated monoglycerides, tristearin, tripalmitin, cetyl esters wax, glyceryl palmitostearate, glyceryl behenate, zein and hydrogenated vegetable oils.
15. An extended release pharmaceutical composition according to claim 12 wherein the composition additionally contain other pharmaceutically acceptable excipients such as fillers, binders, and lubricants.
16. An extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof having a in-vitro dissolution rate when measured using the USP Type II (Paddle apparatus) at 50 rpm in 900 ml/1000 ml, 0.01N hydrochloric acid for first 2 hours followed by the media with pH 6.8 phosphate buffer at 37° C.
from about 5 to about 50% Linezolid released after 1 hour;
from about 10 to about 95% Linezolid released after 4 hours;
from about 35 to about 100% Linezolid released after 8 hours;
from about 55 to about 100% Linezolid released after 12 hours;
from about 70 to about 100% Linezolid released after 16 hours;
and greater than 90% Linezolid released after 24 hours.
17. An extended release pharmaceutical composition according to claim 16 wherein the Linezolid is present from about 50 mg to about 1700 mg.
18. An extended release pharmaceutical composition according to claim 16 wherein the Linezolid is present from about 300 mg to about 1500 mg.
19. An extended release pharmaceutical composition according to claim 16 wherein the Linezolid is present from about 10% w/w to about 95% w/w of the composition.
20. An extended release pharmaceutical composition according to claim 16 wherein the Linezolid is present from about 50% w/w to about 95% w/w of the composition.
21. An extended release pharmaceutical composition according to claim 16 which, upon oral administration to a human, provides a mean maximum plasma concentration (Cmax) of Linezolid not more than about 16 μg/ml.
22. An extended release pharmaceutical composition according to claim 16 which, upon oral administration to a human, provides a mean plasma concentration of at least 2 μg/ml of Linezolid for at least 24 hours.
23. An extended release pharmaceutical composition according to claim 16 wherein pharmaceutical composition comprises of one or more release controlling materials.
24. An extended release pharmaceutical composition according to claim 23 wherein release controlling materials are present from about 0.1% w/w to about 20% w/w of the composition.
25. An extended release pharmaceutical composition according to claim 23 wherein release controlling material is selected from group consisting of hydrophilic release controlling materials or hydrophobic release controlling materials or combinations thereof.
26. An extended release pharmaceutical composition according to claim 25 wherein hydrophilic release controlling material is selected from group consisting of hydroxypropyl methylcellulose, hydroxypropyl cellulose, methylcellulose, povidone, polyethylene glycols, vinyl acetate copolymers, polysaccharides as alginates, xanthan gum, chitosan, carrageenan, dextran, polyalkylene oxides as polyethylene oxide, methaacrylic acid copolymers, maleic anhydride/methyl vinyl ether copolymers, carbomers.
27. An extended release pharmaceutical composition according to claim 25 wherein hydrophobic material is selected from group consisting of polyvinyl acetate dispersion, ethyl cellulose, cellulose acetate, cellulose propionate (lower, medium or higher molecular weight), cellulose acetate propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose triacetate, poly (methyl methacrylate), poly (ethyl methacrylate), poly (butyl methacrylate), poly (isobutyl methacrylate), and poly (hexyl methacrylate), poly (isodecyl methacrylate), poly (lauryl methacrylate), poly (phenyl methacrylate), poly (methyl acrylate), poly (isopropyl acrylate), poly (isobutyl acrylate), poly (octadecyl acrylate), beeswax, carnauba wax, paraffin wax, microcrystalline wax, ozokerite; cetostearyl alcohol, stearyl alcohol, cetyl alcohol and myristyl alcohol, glyceryl monostearate; glycerol monooleate, acetylated monoglycerides, tristearin, tripalmitin, cetyl esters wax, glyceryl palmitostearate, glyceryl behenate, zein and hydrogenated vegetable oils.
28. An extended release pharmaceutical composition according to claim 25 wherein the composition additionally contain other pharmaceutically acceptable excipients such as fillers, binders, and lubricants.
29. An extended release tablet suitable for once daily dosing comprising:
a) 50-1500 mg of Linezolid or pharmaceutically acceptable salt, derivative, prodrug, and metabolite thereof;
b) 0.1% to 20% w/w of one or more release controlling materials;
c) one or more other pharmaceutically acceptable excipients.
30. An extended release pharmaceutical composition according to claim 29 wherein the Linezolid is present from about 10% w/w to about 95% w/w of the composition.
31. An extended release pharmaceutical composition according to claim 29 wherein the Linezolid is present from about 50% w/w to about 95% w/w of the composition.
32. An extended release pharmaceutical composition according to claim 29 which, upon oral administration to a human, provides a mean maximum plasma concentration (Cmax) of Linezolid not more than about 16 μg/ml.
33. An extended release pharmaceutical composition according to claim 29 which, upon oral administration to a human, provides a mean plasma concentration of at least 2 μg/ml of Linezolid for at least 24 hours.
34. An extended release pharmaceutical composition according to claim 29 wherein release controlling material is selected from group consisting of hydrophilic release controlling materials or hydrophobic release controlling materials or combinations thereof.
35. An extended release pharmaceutical composition according to claim 34 wherein hydrophilic release controlling material is selected from group consisting of hydroxypropyl methylcellulose, hydroxypropyl cellulose, methylcellulose, povidone, polyethylene glycols, vinyl acetate copolymers, polysaccharides as alginates, xanthan gum, chitosan, carrageenan, dextran, polyalkylene oxides as polyethylene oxide, methaacrylic acid copolymers, maleic anhydride/methyl vinyl ether copolymers, carbomers.
36. An extended release pharmaceutical composition according to claim 34 wherein hydrophobic material is selected from group consisting of polyvinyl acetate dispersion, ethyl cellulose, cellulose acetate, cellulose propionate (lower, medium or higher molecular weight), cellulose acetate propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose triacetate, poly (methyl methacrylate), poly (ethyl methacrylate), poly (butyl methacrylate), poly (isobutyl methacrylate), and poly (hexyl methacrylate), poly (isodecyl methacrylate), poly (lauryl methacrylate), poly (phenyl methacrylate), poly (methyl acrylate), poly (isopropyl acrylate), poly (isobutyl acrylate), poly (octadecyl acrylate), beeswax, carnauba wax, paraffin wax, microcrystalline wax, ozokerite; cetostearyl alcohol, stearyl alcohol, cetyl alcohol, myristyl alcohol, glyceryl monostearate; glycerol monooleate, acetylated monoglycerides, tristearin, tripalmitin, cetyl esters wax, glyceryl palmitostearate, glyceryl behenate, zein and hydrogenated vegetable oils.
37. An extended release pharmaceutical composition according to claim 34 wherein the composition additionally contain other pharmaceutically acceptable excipients such as fillers, binders, and lubricants.
38. An extended release pharmaceutical composition according to claim 34 wherein the tablet is prepared by direct compression or dry granulation or wet granulation or melt granulation.
39. An extended release pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof and one or more pharmaceutically acceptable excipients so that upon oral administration the maximum concentrations (Cmax) of Linezolid in plasma are statistically significantly lower than the immediate release formulation given twice daily, and area under the plasma concentration-time curve (AUC) and the minimum plasma concentration are maintained over 24 hours.
40. An extended release pharmaceutical composition according to claim 39 wherein the Linezolid is present from about 50 mg to about 1700 mg.
41. An extended release pharmaceutical composition according to claim 39 wherein the Linezolid is present from about 300 mg to about 1500 mg.
42. A method of treating bacterial infections in a mammal comprising administering an extended release, pharmaceutical composition suitable for once daily dosing comprising Linezolid or pharmaceutically acceptable salt, derivative, prodrug, metabolite and polymorph thereof as an active ingredient, and one or more pharmaceutically acceptable excipients, capable of maintaining T>MIC for at least 24 hours.
43. An extended release pharmaceutical composition according to claim 42 wherein the Linezolid is present from about 50 mg to about 1700 mg.
44. An extended release pharmaceutical composition according to claim 42 wherein the Linezolid is present from about 300 mg to about 1500 mg.
US12/980,776 2009-12-29 2010-12-29 Extended release pharmaceutical composition comprising linezolid and process for preparing the same Abandoned US20110159092A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN3193/CHE/2009 2009-12-29
IN3193CH2009 2009-12-29

Publications (1)

Publication Number Publication Date
US20110159092A1 true US20110159092A1 (en) 2011-06-30

Family

ID=43884325

Family Applications (1)

Application Number Title Priority Date Filing Date
US12/980,776 Abandoned US20110159092A1 (en) 2009-12-29 2010-12-29 Extended release pharmaceutical composition comprising linezolid and process for preparing the same

Country Status (2)

Country Link
US (1) US20110159092A1 (en)
WO (1) WO2011080570A2 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103893138A (en) * 2012-12-28 2014-07-02 成都国弘医药有限公司 Tablet containing linezolid crystal form III
US20150246043A1 (en) * 2012-07-27 2015-09-03 Ratiopharm Gmbh Oral dosage forms for modified release comprising ruxolitinib
US10946086B2 (en) 2015-05-04 2021-03-16 Pfizer Inc. Group B Streptococcus polysaccharide-protein conjugates, methods for producing conjugates, immunogenic compositions comprising conjugates, and uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6451345B1 (en) * 2000-01-20 2002-09-17 Eurand Pharmaceuticals Ltd. Functional coating of linezolid microcapsules for taste-masking and associated formulation for oral administration
US6514529B2 (en) * 2000-03-22 2003-02-04 Pharmacia & Upjohn Company Oxazolidinone tablet formulation
US20110300202A1 (en) * 2010-06-02 2011-12-08 Labib Mohamed E Medical item for long term durg release

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040185097A1 (en) * 2003-01-31 2004-09-23 Glenmark Pharmaceuticals Ltd. Controlled release modifying complex and pharmaceutical compositions thereof
US20060068010A1 (en) * 2004-09-30 2006-03-30 Stephen Turner Method for improving the bioavailability of orally delivered therapeutics
PL1749517T3 (en) * 2005-07-20 2008-12-31 Teva Pharma Stable pharmaceutical composition comprising linezolid form IV
EP2144599B1 (en) * 2007-03-02 2010-08-04 Farnam Companies, Inc. Sustained release pellets comprising wax-like material

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6451345B1 (en) * 2000-01-20 2002-09-17 Eurand Pharmaceuticals Ltd. Functional coating of linezolid microcapsules for taste-masking and associated formulation for oral administration
US6514529B2 (en) * 2000-03-22 2003-02-04 Pharmacia & Upjohn Company Oxazolidinone tablet formulation
US20110300202A1 (en) * 2010-06-02 2011-12-08 Labib Mohamed E Medical item for long term durg release

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20150246043A1 (en) * 2012-07-27 2015-09-03 Ratiopharm Gmbh Oral dosage forms for modified release comprising ruxolitinib
CN103893138A (en) * 2012-12-28 2014-07-02 成都国弘医药有限公司 Tablet containing linezolid crystal form III
US10946086B2 (en) 2015-05-04 2021-03-16 Pfizer Inc. Group B Streptococcus polysaccharide-protein conjugates, methods for producing conjugates, immunogenic compositions comprising conjugates, and uses thereof

Also Published As

Publication number Publication date
WO2011080570A3 (en) 2011-11-03
WO2011080570A2 (en) 2011-07-07
WO2011080570A8 (en) 2011-09-09

Similar Documents

Publication Publication Date Title
US8216609B2 (en) Modified release composition of highly soluble drugs
CA2718697C (en) Extended release formulation containing a wax
US8003637B2 (en) Stabilized atypical antipsychotic formulation
US20130280324A1 (en) Sustained release pharmaceutical compositions comprising pregabalin
RU2376988C2 (en) Pharmaceutical composition of slow release, containing aplindor and its derivatives
WO2005048979A2 (en) Pharmaceutical composition having casing with multiple micro tablets
WO2011085188A1 (en) Pharmaceutical compositions comprising anti-psychotic drugs
GB2414668A (en) Sustained release delivery system for tetracycline compounds
US11576917B2 (en) Pharmaceutical compositions comprising Ibrutinib
WO2017208136A1 (en) Pharmaceutical composition of dapagliflozin co-crystal
US20110159092A1 (en) Extended release pharmaceutical composition comprising linezolid and process for preparing the same
EP2867199A2 (en) Stable compositions of fesoterodine
MX2013010598A (en) Controlled release pharmaceutical compositions of selective serotonin reuptake inhibitor.
US20120201886A1 (en) Coated Extended Release Pharmaceutical Compositions Containing Paliperidone
EP3796908B1 (en) Controlled release propiverine formulations
US20100310652A1 (en) Coated extended release pharmaceutical compositions of levetiracetam
US20080081069A1 (en) Novel controlled release formulations of divalproex sodium
US20110195117A1 (en) Controlled release compositions of ropinirole
US20130209553A1 (en) Extended release pharmaceutical compositions of pramipexole
US20200054659A1 (en) Extended release capecitabine capsules
US20140302138A1 (en) Extended release pharmaceutical compositions containing carbamazepine
WO2013057569A2 (en) Extended release pharmaceutical composition containing amoxicillin and clavulanic acid

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION